Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.957
Filtrar
1.
Nature ; 634(8032): 104-112, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39322663

RESUMO

Down syndrome predisposes individuals to haematological abnormalities, such as increased number of erythrocytes and leukaemia in a process that is initiated before birth and is not entirely understood1-3. Here, to understand dysregulated haematopoiesis in Down syndrome, we integrated single-cell transcriptomics of over 1.1 million cells with chromatin accessibility and spatial transcriptomics datasets using human fetal liver and bone marrow samples from 3 fetuses with disomy and 15 fetuses with trisomy. We found that differences in gene expression in Down syndrome were dependent on both cell type and environment. Furthermore, we found multiple lines of evidence that haematopoietic stem cells (HSCs) in Down syndrome are 'primed' to differentiate. We subsequently established a Down syndrome-specific map linking non-coding elements to genes in disomic and trisomic HSCs using 10X multiome data. By integrating this map with genetic variants associated with blood cell counts, we discovered that trisomy restructured regulatory interactions to dysregulate enhancer activity and gene expression critical to erythroid lineage differentiation. Furthermore, as mutations in Down syndrome display a signature of oxidative stress4,5, we validated both increased mitochondrial mass and oxidative stress in Down syndrome, and observed that these mutations preferentially fell into regulatory regions of expressed genes in HSCs. Together, our single-cell, multi-omic resource provides a high-resolution molecular map of fetal haematopoiesis in Down syndrome and indicates significant regulatory restructuring giving rise to co-occurring haematological conditions.


Assuntos
Síndrome de Down , Sangue Fetal , Feto , Hematopoese , Células-Tronco Hematopoéticas , Multiômica , Análise de Célula Única , Humanos , Contagem de Células Sanguíneas , Medula Óssea/metabolismo , Diferenciação Celular/genética , Linhagem da Célula/genética , Cromatina/metabolismo , Cromatina/genética , Síndrome de Down/sangue , Síndrome de Down/embriologia , Síndrome de Down/genética , Síndrome de Down/metabolismo , Síndrome de Down/patologia , Sangue Fetal/citologia , Sangue Fetal/metabolismo , Feto/metabolismo , Feto/citologia , Perfilação da Expressão Gênica , Hematopoese/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Fígado/metabolismo , Fígado/embriologia , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Mutação , Estresse Oxidativo/genética , Reprodutibilidade dos Testes , Transcriptoma/genética , Trissomia/genética
2.
Sci Rep ; 14(1): 21545, 2024 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-39278961

RESUMO

Chronic kidney disease poses a significant threat to public health. Renal replacement therapy is the primary treatment option for end-stage kidney disease. However, there is a promising and relatively new method in regenerative medicine for creating a functional organ known as whole kidney decellularization. This method uses the intrinsic vasculature to perfuse the decellularizing agent into the tissue, effectively penetrating and removing cellular material. The regenerated bioscaffolds could serve as a source of organ donation. This study is focused on evaluating the effectiveness of various SDS exposures in decellularizing human fetal kidneys. The study included human fetal kidneys harvested from fetuses terminated before 14 weeks of gestational age. Kidneys were divided into six treatment groups based on SDS concentration and duration of perfusion. Decellularization, scanning electron microscopy, histopathological staining, immunofluorescent staining, and immunohistochemistry staining were performed to evaluate the adequacy of the process. The statistical analysis revealed that the SDS 0.1% treatment group had the highest collagen deposition after 24 h, significantly greater than the SDS 0.5% treatment group at 24 and 48 h. No significant differences were observed among the other treatment groups. The study concludes that the SDS 0.1% treatment group for 24 h was the most effective in terms of ECM content preservation and effective cell removal. This treatment showed better results than the other treatment groups and can be considered for future whole kidney decellularization studies.


Assuntos
Feto , Rim , Dodecilsulfato de Sódio , Engenharia Tecidual , Humanos , Rim/citologia , Rim/embriologia , Feto/citologia , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Matriz Extracelular/metabolismo , Colágeno/metabolismo , Matriz Extracelular Descelularizada
3.
Stem Cell Res Ther ; 15(1): 303, 2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-39278906

RESUMO

BACKGROUND: Hematopoietic stem progenitor cells (HSPCs) undergo phenotypical and functional changes during their emergence and development. Although the molecular programs governing the development of human hematopoietic stem cells (HSCs) have been investigated broadly, the relationships between dynamic metabolic alterations and their functions remain poorly characterized. METHODS: In this study, we comprehensively described the proteomics of HSPCs in the human fetal liver (FL), umbilical cord blood (UCB), and adult bone marrow (aBM). The metabolic state of human HSPCs was assessed via a Seahorse assay, RT‒PCR, and flow cytometry-based metabolic-related analysis. To investigate whether perturbing glutathione metabolism affects reactive oxygen species (ROS) production, the metabolic state, and the expansion of human HSPCs, HSPCs were treated with buthionine sulfoximine (BSO), an inhibitor of glutathione synthetase, and N-acetyl-L-cysteine (NAC). RESULTS: We investigated the metabolomic landscape of human HSPCs from the fetal, perinatal, and adult developmental stages by in-depth quantitative proteomics and predicted a metabolic switch from the oxidative state to the glycolytic state during human HSPC development. Seahorse assays, mitochondrial activity, ROS level, glucose uptake, and protein synthesis rate analysis supported our findings. In addition, immune-related pathways and antigen presentation were upregulated in UCB or aBM HSPCs, indicating their functional maturation upon development. Glutathione-related metabolic perturbations resulted in distinct responses in human HSPCs and progenitors. Furthermore, the molecular and immunophenotypic differences between human HSPCs at different developmental stages were revealed at the protein level for the first time. CONCLUSION: The metabolic landscape of human HSPCs at three developmental stages (FL, UCB, and aBM), combined with proteomics and functional validations, substantially extends our understanding of HSC metabolic regulation. These findings provide valuable resources for understanding human HSC function and development during fetal and adult life.


Assuntos
Células-Tronco Hematopoéticas , Proteômica , Espécies Reativas de Oxigênio , Humanos , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/citologia , Proteômica/métodos , Espécies Reativas de Oxigênio/metabolismo , Feto/metabolismo , Feto/citologia , Adulto , Sangue Fetal/citologia , Sangue Fetal/metabolismo , Butionina Sulfoximina/farmacologia , Glutationa/metabolismo
4.
Stem Cell Reports ; 19(8): 1189-1204, 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39094562

RESUMO

It has been proposed that adult hematopoiesis is sustained by multipotent progenitors (MPPs) specified during embryogenesis. Adult-like hematopoietic stem cell (HSC) and MPP immunophenotypes are present in the fetus, but knowledge of their functional capacity is incomplete. We found that fetal MPP populations were functionally similar to adult cells, albeit with some differences in lymphoid output. Clonal assessment revealed that lineage biases arose from differences in patterns of single-/bi-lineage differentiation. Long-term (LT)- and short-term (ST)-HSC populations were distinguished from MPPs according to capacity for clonal multilineage differentiation. We discovered that a large cohort of long-term repopulating units (LT-RUs) resides within the ST-HSC population; a significant portion of these were labeled using Flt3-cre. This finding has two implications: (1) use of the CD150+ LT-HSC immunophenotype alone will significantly underestimate the size and diversity of the LT-RU pool and (2) LT-RUs in the ST-HSC population have the attributes required to persist into adulthood.


Assuntos
Linhagem da Célula , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas , Animais , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Camundongos , Diferenciação Celular , Feto/citologia , Imunofenotipagem , Hematopoese , Células Clonais/citologia
5.
Nature ; 632(8025): 603-613, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38987604

RESUMO

A broad range of brain pathologies critically relies on the vasculature, and cerebrovascular disease is a leading cause of death worldwide. However, the cellular and molecular architecture of the human brain vasculature remains incompletely understood1. Here we performed single-cell RNA sequencing analysis of 606,380 freshly isolated endothelial cells, perivascular cells and other tissue-derived cells from 117 samples, from 68 human fetuses and adult patients to construct a molecular atlas of the developing fetal, adult control and diseased human brain vasculature. We identify extensive molecular heterogeneity of the vasculature of healthy fetal and adult human brains and across five vascular-dependent central nervous system (CNS) pathologies, including brain tumours and brain vascular malformations. We identify alteration of arteriovenous differentiation and reactivated fetal as well as conserved dysregulated genes and pathways in the diseased vasculature. Pathological endothelial cells display a loss of CNS-specific properties and reveal an upregulation of MHC class II molecules, indicating atypical features of CNS endothelial cells. Cell-cell interaction analyses predict substantial endothelial-to-perivascular cell ligand-receptor cross-talk, including immune-related and angiogenic pathways, thereby revealing a central role for the endothelium within brain neurovascular unit signalling networks. Our single-cell brain atlas provides insights into the molecular architecture and heterogeneity of the developing, adult/control and diseased human brain vasculature and serves as a powerful reference for future studies.


Assuntos
Neoplasias Encefálicas , Encéfalo , Malformações Vasculares do Sistema Nervoso Central , Células Endoteliais , Feto , RNA-Seq , Análise da Expressão Gênica de Célula Única , Feminino , Humanos , Masculino , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Encéfalo/embriologia , Encéfalo/metabolismo , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/patologia , Comunicação Celular , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Células Endoteliais/citologia , Feto/irrigação sanguínea , Feto/citologia , Feto/embriologia , Malformações Vasculares do Sistema Nervoso Central/patologia , Antígenos HLA-D/metabolismo , Adulto , Saúde
6.
Exp Hematol ; 136: 104585, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39068980

RESUMO

Hematopoietic development goes through a number of embryonic sites that host hematopoietic progenitor and stem cells with function required at specific developmental stages. Among embryonic sites, the fetal liver (FL) hosts definitive hematopoietic stem cells (HSCs) capable of engrafting adult hematopoietic system and supports their rapid expansion. Hence, this site provides an excellent model to understand the cellular and molecular components of the machinery involved in HSC-proliferative events, leading to their overall expansion. It has been unequivocally established that extrinsic regulators orchestrate events that maintain HSC function. Although most studies on extrinsic regulation of HSC function are targeted at adult bone marrow (BM) hematopoiesis, little is known about how FL HSC function is regulated by their microniche. This review provides the current state of our understanding on molecular and cellular niche factors that support FL hematopoiesis.


Assuntos
Células-Tronco Hematopoéticas , Fígado , Nicho de Células-Tronco , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Fígado/citologia , Fígado/embriologia , Animais , Hematopoese , Feto/citologia , Proliferação de Células
7.
Cells ; 13(13)2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38995000

RESUMO

Erythropoiesis occurs first in the yolk sac as a transit "primitive" form, then is gradually replaced by the "definitive" form in the fetal liver (FL) during fetal development and in the bone marrow (BM) postnatally. While it is well known that differences exist between primitive and definitive erythropoiesis, the similarities and differences between FL and BM definitive erythropoiesis have not been studied. Here we performed comprehensive comparisons of erythroid progenitors and precursors at all maturational stages sorted from E16.5 FL and adult BM. We found that FL cells at all maturational stages were larger than their BM counterparts. We further found that FL BFU-E cells divided at a faster rate and underwent more cell divisions than BM BFU-E. Transcriptome comparison revealed that genes with increased expression in FL BFU-Es were enriched in cell division. Interestingly, the expression levels of glucocorticoid receptor Nr3c1, Myc and Myc downstream target Ccna2 were significantly higher in FL BFU-Es, indicating the role of the Nr3c1-Myc-Ccna2 axis in the enhanced proliferation/cell division of FL BFU-E cells. At the CFU-E stage, the expression of genes associated with hemoglobin biosynthesis were much higher in FL CFU-Es, indicating more hemoglobin production. During terminal erythropoiesis, overall temporal patterns in gene expression were conserved between the FL and BM. While biological processes related to translation, the tricarboxylic acid cycle and hypoxia response were upregulated in FL erythroblasts, those related to antiviral signal pathway were upregulated in BM erythroblasts. Our findings uncovered previously unrecognized differences between FL and BM definitive erythropoiesis and provide novel insights into erythropoiesis.


Assuntos
Medula Óssea , Eritropoese , Feto , Fígado , Transcriptoma , Animais , Eritropoese/genética , Camundongos , Fígado/metabolismo , Fígado/embriologia , Fígado/citologia , Transcriptoma/genética , Feto/metabolismo , Feto/citologia , Medula Óssea/metabolismo , Camundongos Endogâmicos C57BL , Regulação da Expressão Gênica no Desenvolvimento , Feminino , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/citologia
8.
Nat Commun ; 15(1): 5898, 2024 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-39003323

RESUMO

Studying human fetal lungs can inform how developmental defects and disease states alter the function of the lungs. Here, we sequenced >150,000 single cells from 19 healthy human pseudoglandular fetal lung tissues ranging between gestational weeks 10-19. We capture dynamic developmental trajectories from progenitor cells that express abundant levels of the cystic fibrosis conductance transmembrane regulator (CFTR). These cells give rise to multiple specialized epithelial cell types. Combined with spatial transcriptomics, we show temporal regulation of key signalling pathways that may drive the temporal and spatial emergence of specialized epithelial cells including ciliated and pulmonary neuroendocrine cells. Finally, we show that human pluripotent stem cell-derived fetal lung models contain CFTR-expressing progenitor cells that capture similar lineage developmental trajectories as identified in the native tissue. Overall, this study provides a comprehensive single-cell atlas of the developing human lung, outlining the temporal and spatial complexities of cell lineage development and benchmarks fetal lung cultures from human pluripotent stem cell differentiations to similar developmental window.


Assuntos
Diferenciação Celular , Regulador de Condutância Transmembrana em Fibrose Cística , Células Epiteliais , Feto , Pulmão , Humanos , Pulmão/embriologia , Pulmão/citologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feto/citologia , Feto/embriologia , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Plasticidade Celular , Linhagem da Célula , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Análise de Célula Única , Transcriptoma , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Transdução de Sinais
9.
Tissue Cell ; 90: 102475, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39059134

RESUMO

Decellularized cortical bone powder derived from adult animals has been shown to induce bone remodeling. Furthermore, it is increasingly evident that the extracellular matrix (ECM) within decellularized tissues differs depending on the source tissue and the age of the animal, leading to distinct effects on cells. In this study, we prepared powders from decellularized fetal and adult porcine bone tissues and conducted biological analyses to determine if the decellularized tissue could induce adipose-derived stem cell differentiation. Decellularized fetal tissues and adult cortical bone were converted into powder by cryomilling, but decellularized adult bone marrow and cartilage were not powdered through this process. In vitro assessments revealed that decellularized fetal tissues, decellularized adult cartilage extract, and decellularized fetal cartilage powder can induce osteoblast differentiation. This study suggests that decellularized fetal bone tissues and adult cartilage contain ECM components that can induce osteoblast differentiation. Additionally, it highlights the utility of decellularized fetal cartilage powder for bone reconstruction.


Assuntos
Cartilagem , Diferenciação Celular , Matriz Extracelular , Feto , Osteogênese , Animais , Cartilagem/citologia , Cartilagem/metabolismo , Matriz Extracelular/metabolismo , Suínos , Feto/citologia , Osso e Ossos/citologia , Osteoblastos/citologia , Osteoblastos/metabolismo , Matriz Extracelular Descelularizada/farmacologia
10.
Stem Cell Res Ther ; 15(1): 235, 2024 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-39075526

RESUMO

BACKGROUND: Hematopoiesis in mammal is a complex and highly regulated process in which hematopoietic stem cells (HSCs) give rise to all types of differentiated blood cells. Previous studies have shown that hairy and enhancer of split (HES) repressors are essential regulators of adult HSC development downstream of Notch signaling. METHODS: In this study, we investigated the role of HES1, a member of HES family, in fetal hematopoiesis using an embryonic hematopoietic specific Hes1 conditional knockout mouse model by using phenotypic flow cytometry, histopathology analysis, and functional in vitro colony forming unit (CFU) assay and in vivo bone marrow transplant (BMT) assay. RESULTS: We found that loss of Hes1 in early embryonic stage leads to smaller embryos and fetal livers, decreases hematopoietic stem progenitor cell (HSPC) pool, results in defective multi-lineage differentiation. Functionally, fetal hematopoietic cells deficient for Hes1 exhibit reduced in vitro progenitor activity and compromised in vivo repopulation capacity in the transplanted recipients. Further analysis shows that fetal hematopoiesis defects in Hes1fl/flFlt3Cre embryos are resulted from decreased proliferation and elevated apoptosis, associated with de-repressed HES1 targets, p27 and PTEN in Hes1-KO fetal HSPCs. Finally, pharmacological inhibition of p27 or PTEN improves fetal HSPCs function both in vitro and in vivo. CONCLUSION: Together, our findings reveal a previously unappreciated role for HES1 in regulating fetal hematopoiesis, and provide new insight into the differences between fetal and adult HSC maintenance.


Assuntos
Feto , Hematopoese , Células-Tronco Hematopoéticas , Camundongos Knockout , Fatores de Transcrição HES-1 , Animais , Fatores de Transcrição HES-1/metabolismo , Fatores de Transcrição HES-1/genética , Camundongos , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/citologia , Feto/citologia , Feto/metabolismo , Diferenciação Celular , Apoptose , Proliferação de Células , PTEN Fosfo-Hidrolase/metabolismo , PTEN Fosfo-Hidrolase/genética , Transdução de Sinais , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/genética
11.
Life Sci Alliance ; 7(8)2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38830768

RESUMO

Hematopoietic stem cells and multipotential progenitors emerge in multiple, overlapping waves of fetal development. Some of these populations seed the bone marrow and sustain adult B- and T-cell development long-term after birth. However, others are present transiently, but whether they are vestigial or generate B and T cells that contribute to the adult immune system is not well understood. We now report that transient fetal progenitors distinguished by expression of low levels of the PU.1 transcription factor generated activated and memory T and B cells that colonized and were maintained in secondary lymphoid tissues. These included the small and large intestines, where they may contribute to the maintenance of gut homeostasis through at least middle age. At least some of the activated/memory cells may have been the progeny of B-1 and marginal zone B cells, as transient PU.1low fetal progenitors efficiently generated those populations. Taken together, our data demonstrate the potential of B- and T-cell progeny of transient PU.1low fetal progenitors to make an early and long-term contribution to the adult immune system.


Assuntos
Linfócitos B , Proteínas Proto-Oncogênicas , Linfócitos T , Transativadores , Transativadores/metabolismo , Transativadores/genética , Animais , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/genética , Camundongos , Linfócitos B/metabolismo , Linfócitos B/imunologia , Linfócitos B/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/citologia , Camundongos Endogâmicos C57BL , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/citologia , Diferenciação Celular/imunologia , Feminino , Feto/citologia , Células-Tronco Fetais/metabolismo , Células-Tronco Fetais/citologia
12.
Int J Mol Sci ; 25(9)2024 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-38732014

RESUMO

Fetal organs and organoids are important tools for studying organ development. Recently, porcine organs have garnered attention as potential organs for xenotransplantation because of their high degree of similarity to human organs. However, to meet the prompt demand for porcine fetal organs by patients and researchers, effective methods for producing, retrieving, and cryopreserving pig fetuses are indispensable. Therefore, in this study, to collect fetuses for kidney extraction, we employed cesarean sections to preserve the survival and fertility of the mother pig and a method for storing fetal kidneys by long-term cryopreservation. Subsequently, we evaluated the utility of these two methods. We confirmed that the kidneys of pig fetuses retrieved by cesarean section that were cryopreserved for an extended period could resume renal growth when grafted into mice and were capable of forming renal organoids. These results demonstrate the usefulness of long-term cryopreserved fetal pig organs and strongly suggest the effectiveness of our comprehensive system of pig fetus retrieval and fetal organ preservation, thereby highlighting its potential as an accelerator of xenotransplantation research and clinical innovation.


Assuntos
Criopreservação , Feto , Transplante de Rim , Rim , Organoides , Animais , Criopreservação/métodos , Suínos , Rim/citologia , Organoides/citologia , Organoides/transplante , Camundongos , Transplante de Rim/métodos , Feto/citologia , Feminino , Transplante Heterólogo/métodos , Preservação de Órgãos/métodos
13.
Cells ; 13(9)2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38727278

RESUMO

Spermatogenesis involves a complex process of cellular differentiation maintained by spermatogonial stem cells (SSCs). Being critical to male reproduction, it is generally assumed that spermatogenesis starts and ends in equivalent transcriptional states in related species. Based on single-cell gene expression profiling, it has been proposed that undifferentiated human spermatogonia can be subclassified into four heterogenous subtypes, termed states 0, 0A, 0B, and 1. To increase the resolution of the undifferentiated compartment and trace the origin of the spermatogenic trajectory, we re-analysed the single-cell (sc) RNA-sequencing libraries of 34 post-pubescent human testes to generate an integrated atlas of germ cell differentiation. We then used this atlas to perform comparative analyses of the putative SSC transcriptome both across human development (using 28 foetal and pre-pubertal scRNA-seq libraries) and across species (including data from sheep, pig, buffalo, rhesus and cynomolgus macaque, rat, and mouse). Alongside its detailed characterisation, we show that the transcriptional heterogeneity of the undifferentiated spermatogonial cell compartment varies not only between species but across development. Our findings associate 'state 0B' with a suppressive transcriptomic programme that, in adult humans, acts to functionally oppose proliferation and maintain cells in a ready-to-react state. Consistent with this conclusion, we show that human foetal germ cells-which are mitotically arrested-can be characterised solely as state 0B. While germ cells with a state 0B signature are also present in foetal mice (and are likely conserved at this stage throughout mammals), they are not maintained into adulthood. We conjecture that in rodents, the foetal-like state 0B differentiates at birth into the renewing SSC population, whereas in humans it is maintained as a reserve population, supporting testicular homeostasis over a longer reproductive lifespan while reducing mutagenic load. Together, these results suggest that SSCs adopt differing evolutionary strategies across species to ensure fertility and genome integrity over vastly differing life histories and reproductive timeframes.


Assuntos
Espermatogônias , Humanos , Animais , Masculino , Espermatogônias/citologia , Espermatogônias/metabolismo , Células-Tronco Germinativas Adultas/metabolismo , Células-Tronco Germinativas Adultas/citologia , Diferenciação Celular/genética , Espermatogênese/genética , Transcriptoma/genética , Adulto , Camundongos , Feto/citologia , Testículo/citologia , Testículo/metabolismo , Roedores , Ratos , Análise de Célula Única
14.
Mol Neurobiol ; 61(8): 5738-5753, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38227271

RESUMO

Oligodendrocytes (OL) are the myelinating cells of the central nervous system that mediate nerve conduction. Loss of oligodendrocytes results in demyelination, triggering neurological deficits. Developing a better understanding of the cell signaling pathways influencing OL development may aid in the development of therapeutic strategies. The primary focus of this study was to investigate and elucidate the cell signaling pathways implicated in the developmental maturation of oligodendrocytes using human fetal neural stem cells (hFNSCs)-derived primary OL and MO3.13 cell line. Successful differentiation into OL was established by examining morphological changes, increased expression of mature OL markers MBP, MOG and decreased expression of pre-OL markers CSPG4 and O4. Analyzing transcriptional datasets (using RNA sequencing) in pre-OL and mature OL derived from hFNSCs revealed the novel and critical involvement of the JAK-STAT cell signaling pathway in terminal OL maturation. The finding was validated in MO3.13 cell line whose differentiation was accompanied by upregulation of IL-6 and the transcription factor STAT3. Increased phosphorylated STAT3 (pY705) levels were demonstrated by western blotting in hFNSCs-derived primary OL as well as terminal maturation in MO3.13 cells, thus validating the involvement of the JAK-STAT pathway in OL maturation. Pharmacological suppression of STAT3 phosphorylation (confirmed by western blotting) was able to prevent the increase of MBP-positive cells as demonstrated by flow cytometry. These novel findings highlight the involvement of the JAK-STAT pathway in OL maturation and raise the possibility of using this as a therapeutic strategy in demyelinating diseases.


Assuntos
Diferenciação Celular , Janus Quinases , Células-Tronco Neurais , Oligodendroglia , Fator de Transcrição STAT3 , Transdução de Sinais , Humanos , Oligodendroglia/metabolismo , Oligodendroglia/citologia , Transdução de Sinais/fisiologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/citologia , Diferenciação Celular/fisiologia , Janus Quinases/metabolismo , Fator de Transcrição STAT3/metabolismo , Fatores de Transcrição STAT/metabolismo , Fosforilação , Linhagem Celular , Feto/citologia , Células-Tronco Fetais/metabolismo , Interleucina-6/metabolismo
15.
Nature ; 625(7996): 788-796, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38029793

RESUMO

The expansion of the neocortex, a hallmark of mammalian evolution1,2, was accompanied by an increase in cerebellar neuron numbers3. However, little is known about the evolution of the cellular programmes underlying the development of the cerebellum in mammals. In this study we generated single-nucleus RNA-sequencing data for around 400,000 cells to trace the development of the cerebellum from early neurogenesis to adulthood in human, mouse and the marsupial opossum. We established a consensus classification of the cellular diversity in the developing mammalian cerebellum and validated it by spatial mapping in the fetal human cerebellum. Our cross-species analyses revealed largely conserved developmental dynamics of cell-type generation, except for Purkinje cells, for which we observed an expansion of early-born subtypes in the human lineage. Global transcriptome profiles, conserved cell-state markers and gene-expression trajectories across neuronal differentiation show that cerebellar cell-type-defining programmes have been overall preserved for at least 160 million years. However, we also identified many orthologous genes that gained or lost expression in cerebellar neural cell types in one of the species or evolved new expression trajectories during neuronal differentiation, indicating widespread gene repurposing at the cell-type level. In sum, our study unveils shared and lineage-specific gene-expression programmes governing the development of cerebellar cells and expands our understanding of mammalian brain evolution.


Assuntos
Cerebelo , Evolução Molecular , Mamíferos , Neurogênese , Animais , Humanos , Camundongos , Linhagem da Célula/genética , Cerebelo/citologia , Cerebelo/embriologia , Cerebelo/crescimento & desenvolvimento , Feto/citologia , Feto/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Neurogênese/genética , Neurônios/citologia , Neurônios/metabolismo , Gambás/embriologia , Gambás/crescimento & desenvolvimento , Células de Purkinje/citologia , Células de Purkinje/metabolismo , Análise da Expressão Gênica de Célula Única , Especificidade da Espécie , Transcriptoma , Mamíferos/embriologia , Mamíferos/crescimento & desenvolvimento
16.
Science ; 381(6664): 1286, 2023 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-37733842

RESUMO

Shifting pools of antigen can influence pregnancy-induced immune tolerance.


Assuntos
Quimerismo , Feto , Tolerância Imunológica , Troca Materno-Fetal , Criança , Feminino , Humanos , Gravidez , Troca Materno-Fetal/imunologia , Antígenos/imunologia , Feto/citologia , Feto/imunologia
17.
Science ; 381(6664): 1324-1330, 2023 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-37733857

RESUMO

Pregnancy confers partner-specific protection against complications in future pregnancy that parallel persistence of fetal microchimeric cells (FMcs) in mothers after parturition. We show that preexisting FMcs become displaced by new FMcs during pregnancy and that FMc tonic stimulation is essential for expansion of protective fetal-specific forkhead box P3 (FOXP3)-positive regulatory T cells (Treg cells). Maternal microchimeric cells and accumulation of Treg cells with noninherited maternal antigen (NIMA) specificity are similarly overturned in daughters after pregnancy, highlighting a fixed microchimeric cell niche. Whereas NIMA-specific tolerance is functionally erased by pregnancy, partner-specific resiliency against pregnancy complications persists in mothers despite paternity changes in intervening pregnancy. Persistent fetal tolerance reflects FOXP3 expression plasticity, which allows mothers to more durably remember their babies, whereas daughters forget their mothers with new pregnancy-imprinted immunological memories.


Assuntos
Quimerismo , Feto , Tolerância Imunológica , Memória Imunológica , Troca Materno-Fetal , Gravidez , Animais , Feminino , Camundongos , Gravidez/imunologia , Antígenos/imunologia , Plasticidade Celular , Feto/citologia , Feto/imunologia , Fatores de Transcrição Forkhead/imunologia , Troca Materno-Fetal/imunologia , Camundongos Endogâmicos C57BL , Linfócitos T Reguladores/imunologia
19.
Am J Med Genet B Neuropsychiatr Genet ; 192(3-4): 62-70, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36863698

RESUMO

Investigating functional, temporal, and cell-type expression features of mutations is important for understanding a complex disease. Here, we collected and analyzed common variants and de novo mutations (DNMs) in schizophrenia (SCZ). We collected 2,636 missense and loss-of-function (LoF) DNMs in 2,263 genes across 3,477 SCZ patients (SCZ-DNMs). We curated three gene lists: (a) SCZ-neuroGenes (159 genes), which are intolerant to LoF and missense DNMs and are neurologically important, (b) SCZ-moduleGenes (52 genes), which were derived from network analyses of SCZ-DNMs, and (c) SCZ-commonGenes (120 genes) from a recent GWAS as reference. To compare temporal gene expression, we used the BrainSpan dataset. We defined a fetal effect score (FES) to quantify the involvement of each gene in prenatal brain development. We further employed the specificity indexes (SIs) to evaluate cell-type expression specificity from single-cell expression data in cerebral cortices of humans and mice. Compared with SCZ-commonGenes, SCZ-neuroGenes and SCZ-moduleGenes were highly expressed in the prenatal stage, had higher FESs, and had higher SIs in fetal replicating cells and undifferentiated cell types. Our results suggested that gene expression patterns in specific cell types in early fetal stages might have impacts on the risk of SCZ during adulthood.


Assuntos
Encéfalo , Mutação , Esquizofrenia , Esquizofrenia/genética , Esquizofrenia/patologia , Esquizofrenia/fisiopatologia , Encéfalo/citologia , Encéfalo/embriologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/patologia , Animais , Camundongos , Feto/citologia , Feto/embriologia , Neurônios/metabolismo , Mutação com Perda de Função , Mutação de Sentido Incorreto , Humanos , Especificidade de Órgãos
20.
Mol Psychiatry ; 28(4): 1571-1584, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36385168

RESUMO

Prenatal alcohol exposure is the foremost preventable etiology of intellectual disability and leads to a collection of diagnoses known as Fetal Alcohol Spectrum Disorders (FASD). Alcohol (EtOH) impacts diverse neural cell types and activity, but the precise functional pathophysiological effects on the human fetal cerebral cortex are unclear. Here, we used human cortical organoids to study the effects of EtOH on neurogenesis and validated our findings in primary human fetal neurons. EtOH exposure produced temporally dependent cellular effects on proliferation, cell cycle, and apoptosis. In addition, we identified EtOH-induced alterations in post-translational histone modifications and chromatin accessibility, leading to impairment of cAMP and calcium signaling, glutamatergic synaptic development, and astrocytic function. Proteomic spatial profiling of cortical organoids showed region-specific, EtOH-induced alterations linked to changes in cytoskeleton, gliogenesis, and impaired synaptogenesis. Finally, multi-electrode array electrophysiology recordings confirmed the deleterious impact of EtOH on neural network formation and activity in cortical organoids, which was validated in primary human fetal tissues. Our findings demonstrate progress in defining the human molecular and cellular phenotypic signatures of prenatal alcohol exposure on functional neurodevelopment, increasing our knowledge for potential therapeutic interventions targeting FASD symptoms.


Assuntos
Córtex Cerebral , Etanol , Vias Neurais , Neurogênese , Neurônios , Organoides , Feminino , Humanos , Masculino , Gravidez , Astrócitos/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Córtex Cerebral/citologia , Montagem e Desmontagem da Cromatina/efeitos dos fármacos , Montagem e Desmontagem da Cromatina/genética , Epigênese Genética/efeitos dos fármacos , Epigênese Genética/genética , Etanol/farmacologia , Transtornos do Espectro Alcoólico Fetal/etiologia , Transtornos do Espectro Alcoólico Fetal/genética , Feto/citologia , Perfilação da Expressão Gênica , Rede Nervosa/efeitos dos fármacos , Transtornos do Neurodesenvolvimento/induzido quimicamente , Transtornos do Neurodesenvolvimento/genética , Transtornos do Neurodesenvolvimento/patologia , Neurogênese/efeitos dos fármacos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Organoides/citologia , Organoides/efeitos dos fármacos , Organoides/patologia , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/genética , Proteômica , Sinapses/efeitos dos fármacos , Vias Neurais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA